42 results on '"Heath, William R."'
Search Results
2. Intrahepatic activation of naive CD4+ T cells by liver-resident phagocytic cells
- Author
-
Tay, Szun, Wong, Yik Chun, Roediger, Ben, Sierro, Frederic, Lu, Bo, McDonald, David M., McGuffog, Claire M., Meyer, Nicholas J., Alexander, Ian E., Parish, Ian, Heath, William R., Tay, Szun, Wong, Yik Chun, Roediger, Ben, Sierro, Frederic, Lu, Bo, McDonald, David M., McGuffog, Claire M., Meyer, Nicholas J., Alexander, Ian E., Parish, Ian, and Heath, William R.
- Abstract
Naive T cell activation is normally restricted to the lymphoid organs, in part because of their limited ability to migrate into the parenchyma of peripheral tissues. The liver vasculature is unique, however, and circulating leukocytes within the hepatic sinusoids have direct access to liver-resident cells, which include an abundant population of Kupffer cells. It is well accepted that recognition of cognate Ag within the liver leads to naive CD8+T cell activation in situ, but it is unclear whether the liver also supports naive CD4+T cell activation. In this study, we show that naive CD4+T cells can be activated to proliferate in the liver when cognate Ag expression is induced in hepatocytes by recombinant adeno-associated viral vectors. Ag-specific retention and activation of naive CD4+T cells within the liver are independent of lymphoid tissues but dependent on a clodronate liposome-sensitive population of liver-resident phagocytic cells. To our knowledge, this study provides the first unequivocal evidence that naive CD4+T cells can be activated in a nonlymphoid organ. It also gives critical insight into how CD4+T cells specific for Ag expressed in the liver are recruited to participate in protective or pathological responses during hepatotropic infections and autoimmune liver disease.
- Published
- 2014
3. Breakdown in repression of IFN-y mRNA leads to accumulation of self-reactive effector CD8+ T cells
- Author
-
Chang, Pheh-Ping, Lee, Sau K, Hu, Xin, Davey, Gayle, Duan, Guowen, Cho, Jae-Ho, Karupiah, Guna, Sprent, Jonathan, Heath, William R, Bertram, Edward M, Vinuesa, Carola, Chang, Pheh-Ping, Lee, Sau K, Hu, Xin, Davey, Gayle, Duan, Guowen, Cho, Jae-Ho, Karupiah, Guna, Sprent, Jonathan, Heath, William R, Bertram, Edward M, and Vinuesa, Carola
- Abstract
Tight regulation of virus-induced cytotoxic effector CD8+ T cells is essential to prevent immunopathology. Naturally occurring effector CD8+ T cells, with a KLRG1hi CD62Llo phenotype typical of short-lived effector CD8+ T cells (SLECs), can be found in increased numbers in autoimmune-prone mice, most notably in mice homozygous for the san allele of Roquin. These SLEC-like cells were able to trigger autoimmune diabetes in a susceptible background. When Roquin is mutated (Roquinsan), effector CD8+ T cells accumulate in a cell-autonomous manner, most prominently as SLEC-like effectors. Excessive IFN-y promotes the accumulation of SLEC-like cells, increases their T-bet expression, and enhances their granzyme B production in vivo. We show that overexpression of IFN-y was caused by failed posttranscriptional repression of Ifng mRNA. This study identifies a novel mechanism that prevents accumulation of self-reactive cytotoxic effectors, highlighting the importance of regulating Ifng mRNA stability to maintain CD8+ T cell homeostasis and prevent CD8-mediated autoimmunity.
- Published
- 2012
4. T Cell Receptor Antagonist Peptides Induce Positive Selection.
- Author
-
Hogquist, Kristin A., Jameson, Stephen C., Heath, William R., Howard, Jane L., Bevan, Michael J., and Carbone, Francis R.
- Subjects
- *
T cell receptors , *PEPTIDES , *ORGAN culture , *FETUS , *THYMUS , *TRANSGENIC mice , *LABORATORY mice - Abstract
We have used organ culture of fetal thymic lobes from T cell receptor (TCR) transgenic &brg;2M(-/-) mice to study the role of peptides in positive selection. The TCR used was from a CD8+ T cell specific for ovalbumin 257-264 in the context of Kb. Several peptides with the ability to induce positive selection were identified. These pep-tide-selected thymocytes have the same phenotype as mature CD8+ T cells and can respond to antigen. Those peptides with the ability to induce positive se-lection were all variants of the antigenic peptide and were identified as TCR antagonist peptides for this re-ceptor. One peptide tested, E1, induced positive selec-tion on the &brg;2M[-l-) background but negative selec-tion on the &brg;2M(+l-) background. These results show that the process of positive selection is exquisitely peptide specific and sensitive to extremely low ligand density and support the notion that low efficacy li-gands mediate positive selection. [ABSTRACT FROM AUTHOR]
- Published
- 2012
5. Classical Type 1 Dendritic Cells Dominate Priming of Th1 Responses to Herpes Simplex Virus Type 1 Skin Infection.
- Author
-
Harpur, Christopher M., Yu Kato, Dewi, Shinta T., Stankovic, Sanda, Johnson, Darryl N., Bedoui, Sammy, Whitney, Paul G., Lahoud, Mireille H., Caminschi, Irina, Heath, William R., Brooks, Andrew G., and Gebhardt, Thomas
- Subjects
- *
HUMAN herpesvirus 1 , *DENDRITIC cells - Abstract
CD4+ T cell responses are crucial for the control of many intracellular pathogens, yet the requirements for their induction are not fully understood. To better understand the role that various dendritic cell (DC) subtypes play in CD4+ T cell priming, we compared in vivo T cell responses to skin inoculation of mice with infectious or UV-inactivated HSV type 1. Localized infection elicited a Th1 response that was primed in skin-draining lymph nodes involving Ag presentation by migratory dermal and lymph node-resident DC. However, expansion and Th1 differentiation was impaired in response to UV-inactivated virus (UV-HSV), and this defect correlated with a restriction of Ag presentation to migratory CD103- dermal DC. A similar differentiation defect was seen in infected mice lacking CD8α+ and CD103+ classical type 1 DC (cDC1). Finally, Th1 differentiation after UV-HSV inoculation was rescued by targeted Ag delivery to CD8α+ and CD103+ cDC1 using an anti-Clec9A Ab construct. This suggests that Ag presentation by cDC1 is crucial for optimal Th1 immunity to HSV type 1 infection and potentially other pathogens of the skin. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
6. IFN Regulatory Factor 3 Balances Th1 and T Follicular Helper Immunity during Nonlethal Blood-Stage Plasmodium Infection.
- Author
-
Liligeto, Urijah N., Nair, Arya Sheela, Fogg, Lily G., Best, Shannon E., Lansink, Lianne I. M., James, Kylie R., Soon, Megan S. F., Sebina, Ismail, Edwards, Chelsea L., Haque, Ashraful, Fernandez-Ruiz, Daniel, Davey, Gayle, Heath, William R., Schroder, Kate, Engwerda, Christian R., Wilson, Jane A. C., Suhrbier, Andreas, Austin, Rebecca, Lane, Steven W., and Hill, Geoffrey R.
- Subjects
- *
INTERFERON regulatory factor genetics , *T helper cells , *MALARIA immunology , *CD4 antigen , *CELL differentiation , *PLASMODIUM - Abstract
Differentiation of CD4+ Th cells is critical for immunity to malaria. Several innate immune signaling pathways have been implicated in the detection of blood-stage Plasmodium parasites, yet their influence over Th cell immunity remains unclear. In this study, we used Plasmodium-reactive TCR transgenic CD4+ T cells, termed PbTII cells, during nonlethal P. chabaudi chabaudi AS and P. yoelii 17XNL infection in mice, to examine Th cell development in vivo. We found no role for caspase1/11, stimulator of IFN genes, or mitochondrial antiviral-signaling protein, and only modest roles for MyD88 and TRIF-dependent signaling in controlling PbTII cell expansion. In contrast, IFN regulatory factor 3 (IRF3) was important for supporting PbTII expansion, promoting Th1 over T follicular helper (Tfh) differentiation, and controlling parasites during the first week of infection. IRF3 was not required for early priming by conventional dendritic cells, but was essential for promoting CXCL9 and MHC class II expression by inflammatory monocytes that supported PbTII responses in the spleen. Thereafter, IRF3-deficiency boosted Tfh responses, germinal center B cell and memory B cell development, parasite-specific Ab production, and resolution of infection. We also noted a B cell-intrinsic role for IRF3 in regulating humoral immune responses. Thus, we revealed roles for IRF3 in balancing Th1- and Tfh-dependent immunity during nonlethal infection with blood-stage Plasmodium parasites. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
7. Chemokine Receptor-Dependent Control of Skin Tissue-Resident Memory T Cell Formation.
- Author
-
Zaid, Ali, Jyh Liang Hor, Christo, Susan N., Groom, Joanna R., Heath, William R., Mackay, Laura K., and Mueller, Scott N.
- Subjects
- *
CHEMOKINE receptors , *SKIN , *T cells , *KERATINOCYTES , *ACTOMYOSIN - Abstract
Infection or inflammation of the skin recruits effector CD8+ T cells that enter the epidermis and form populations of long-lived tissue-resident memory T (TRM) cells. These skin TRM cells migrate within the constrained epidermal environment by extending multiple dynamic dendritic projections and squeezing between keratinocytes to survey the tissue for pathogens. In this study, we examined the signals required for this distinctive mode of T cell migration by inhibiting key cytoskeletal components and performing intravital two-photon microscopy to visualize TRM cell behavior. We found that TRM cell motility and dendrite formation required an intact actomyosin cytoskeleton and the Rho-associated coiled-coil containing kinases. We also identified an essential role for microtubules for maintaining skin TRM cell shape and cellular integrity. We reveal a role for pertussis toxinsensitive signaling for TRM cell dendritic morphology and migration that is independent of CXCR3 or CXCR6, or the skinselective chemokine receptors CCR10 and CCR8. However, we found that CXCR6 and CCR10 expression by CD8+ T cells was required for the optimal formation of memory T cell populations, in particular TRM cell populations in the skin. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
8. Targeting Antigen to Clec9A Primes Follicular Th Cell Memory Responses Capable of Robust Recall.
- Author
-
Yu Kato, Zaid, Ali, Davey, Gayle M., Mueller, Scott N., Nutt, Stephen L., Zotos, Dimitra, Tarlinton, David M., Shortman, Ken, Lahoud, Mireille H., Heath, William R., and Caminschi, Irina
- Subjects
- *
T helper cells , *IMMUNOREGULATION , *F-actin , *GENE targeting , *HUMORAL immunity , *CD4 antigen - Abstract
Targeting Ags to dendritic cell (DC) surface receptors can induce a variety of responses depending on the DC type targeted, the receptor targeted, and the adjuvant used. Clec9A (DNGR-1), which is expressed by CD8+ DCs, has been shown to bind F-actin exposed on damaged cells. Targeting Ag to this receptor in mice and nonhuman primates induces strong humoral immunity even in the absence of adjuvant, a process seen for a few select DC receptors. In contrast with other receptors, however, targeting Clec9A induces long-lived, affinity-matured Ab responses that are associated with efficient CD4+ T cell responses shown to possess properties of follicular Th cells (TFH). In this article, we provide definitive evidence that Clec9A targeting promotes the development of TFH by showing that responding CD4 T cells express CXCR5, PD1, the TFH transcription factor Bcl6, and the cytokine IL-21, and that these cells localize to germinal centers. Furthermore, we extend studies from the model Ag OVA to the viral Ag glycoprotein D of HSV-1 and examine the capacity of primed TFH to form functional memory. We show that targeting glycoprotein D to Clec9A even in the absence of adjuvant induced long-lived memory CXCR5+ PD1hi CD4+ T cells that proliferated extensively upon secondary challenge and rapidly developed into effector TFH. This was associated with enhanced germinal center B cell responses and accelerated Ab production. Our study indicates that targeting Ags to Clec9A in the absence of adjuvant routinely generates TFH responses that form long-lived memory capable of robust secondary TFH responses. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
9. Intrahepatic Activation of Naive CD4+ T Cells by Liver-Resident Phagocytic Cells.
- Author
-
Tay, Szun S., Yik Chun Wong, Roediger, Ben, Sierro, Frederic, Lu, Bo, McDonald, David M., McGuffog, Claire M., Meyer, Nicholas J., Alexander, Ian E., Parish, Ian A., Heath, William R., Weninger, Wolfgang, Bishop, G. Alex, Gamble, Jennifer R., McCaughan, Geoffrey W., Bertolino, Patrick, and Bowen, David G.
- Subjects
- *
T cells , *CD4 antigen , *PHAGOCYTES , *KUPFFER cells , *CD8 antigen , *LIVER physiology , *AUTOIMMUNE diseases , *PHYSIOLOGY - Abstract
Naive T cell activation is normally restricted to the lymphoid organs, in part because of their limited ability to migrate into the parenchyma of peripheral tissues. The liver vasculature is unique, however, and circulating leukocytes within the hepatic sinusoids have direct access to liver-resident cells, which include an abundant population of Kupffer cells. It is well accepted that recognition of cognate Ag within the liver leads to naive CD8+ T cell activation in situ, but it is unclear whether the liver also supports naive CD4+ T cell activation. In this study, we show that naive CD4+ T cells can be activated to proliferate in the liver when cognate Ag expression is induced in hepatocytes by recombinant adeno-associated viral vectors. Ag-specific retention and activation of naive CD4+ T cells within the liver are independent of lymphoid tissues but dependent on a clodronate liposome-sensitive population of liver-resident phagocytic cells. To our knowledge, this study provides the first unequivocal evidence that naive CD4+ T cells can be activated in a nonlymphoid organ. It also gives critical insight into how CD4+ T cells specific for Ag expressed in the liver are recruited to participate in protective or pathological responses during hepatotropic infections and autoimmune liver disease. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
10. Breakdown in Repression of IFN-γ mRNA Leads to Accumulation of Self-Reactive Effector CD8+ Τ Cells.
- Author
-
Pheh-Ping Chang, Lee, Sau Κ., Xin Hu, Davey, Gayle, Guowen Duan, Jae-Ho Cho, Karupiah, Guna, Sprent, Jonathan, Heath, William R., Bertram, Edward M., and Vinuesa, Carola G.
- Subjects
- *
MESSENGER RNA , *IMMUNOPATHOLOGY , *DIABETES , *GRANZYMES , *AUTOIMMUNITY , *LABORATORY mice - Abstract
Tight regulation of virus-induced cytotoxic effector CD8+ Τ cells is essential to prevent immunopathology. Naturally occurring effector CD8+ Τ cells, with a KLRG1hi CD62Llo phenotype typical of short-lived effector CD8+ Τ cells (SLECs), can be found in increased numbers in autoimmune-prone mice, most notably in mice homozygous for the san allele of Roquin. These SLEC-like cells were able to trigger autoimmune diabetes in a susceptible background. When Roquin is mutated (Roquinsan), effector CD8+ Τ cells accumulate in a cell-autonomous manner, most prominently as SLEC-like effectors. Excessive IFN-γ promotes the accumulation of SLEC-like cells, increases their T-bet expression, and enhances their granzyme Β production in vivo. We show that overexpression of IFN-γ was caused by failed posttranscriptional repression of Ifng mRNA. This study identifies a novel mechanism that prevents accumulation of self-reactive cytotoxic effectors, highlighting the importance of regulating Ifng mRNA stability to maintain CD8+ Τ cell homeostasis and prevent CD8-mediated autoimmunity. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
11. Maintenance of T Cell Function in the Face of Chronic Antigen Stimulation and Repeated Reactivation for a Latent Virus Infection.
- Author
-
Mackay, Laura K., Wakim, Linda, van Vliet, Catherine J., Jones, Claerwen M., Mueller, Scott N., Bannard, Oliver, Fearon, Douglas T., Heath, William R., and Carbone, Francis R.
- Subjects
- *
T cells , *CELL physiology , *LATENT infection , *PATHOGENIC microorganisms , *HERPESVIRUSES , *CELL proliferation , *CYTOKINES - Abstract
Persisting infections are often associated with chronic T cell activation. For certain pathogens, this can lead to T cell exhaustion and survival of what is otherwise a cleared infection. In contrast, for herpesviruses, T cells never eliminate infection once it is established. Instead, effective immunity appears to maintain these pathogens in a state of latency. We used infection with HSV to examine whether effector-type T cells undergoing chronic stimulation retained functional and proliferative capacity during latency and subsequent reactivation. We found that latency-associated T cells exhibited a polyfunctional phenotype and could secrete a range of effector cytokines. These T cells were also capable of mounting a recall proliferative response on HSV reactivation and could do so repeatedly. Thus, for this latent infection, T cells subjected to chronic Ag stimulation and periodic reactivation retain the ability to respond to local virus challenge. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
12. Targeting Antigen to Mouse Dendritic Cells via Clec9A Induces Potent CD4 T Cell Responses Biased toward a Follicular Helper Phenotype.
- Author
-
Lahoud, Mireille H., Ahmet, Fatma, Kitsoulis, Susie, Soo San Wan, Vremec, David, Chin-Nien Lee, Phipson, Belinda, Wei Shi, Smyth, Gordon K., Lew, Andrew M., Yu Kato, Mueller, Scott N., Davey, Gayle M., Heath, William R., Shortman, Ken, and Caminschi, Irina
- Subjects
- *
ANTIGENS , *DENDRITIC cells , *IMMUNOLOGICAL adjuvants , *CELL proliferation , *T cells - Abstract
Three surface molecules of mouse CD8+ dendritic cells (DCs), also found on the equivalent human DC subpopulation, were compared as targets for Ab-mediated delivery of Ags, a developing strategy for vaccination. For the production of cytotoxic T cells, DEC-205 and Clec9A, but not Clec12A, were effective targets, although only in the presence of adjuvants. For Ab production, however, Clec9A excelled as a target, even in the absence of adjuvant. Potent humoral immunity was a result of the highly specific expression of Clec9A on DCs, which allowed longer residence of targeting Abs in the bloodstream, prolonged DC Ag presentation, and extended CD4 T cell proliferation, all of which drove highly efficient development of follicular helper T cells. Because Clec9A shows a similar expression pattern on human DCs, it has particular promise as a target for vaccines of human application. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
13. Granzyme B Expression by CD8+ T Cells Is Required for the Development of Experimental Cerebral Malaria.
- Author
-
Haque, Ashraful, Best, Shannon E., Unosson, Klara, Amante, Fiona H., de Labastida, Fabian, Anstey, Nicholas M., Karupiah, Gunasegaran, Smyth, Mark J., Heath, William R., and Engwerda, Christian R.
- Subjects
- *
MALARIA , *PARASITES , *T cells , *ANTIPARASITIC agents , *BRAIN diseases , *PHYSIOLOGY , *DISEASE risk factors - Abstract
Parasite burden predicts disease severity in malaria and risk of death in cerebral malaria patients. In murine experimental cerebral malaria (ECM), parasite burden and CD8+ T cells promote disease by mechanisms that are not fully understood. We found that the majority of brain-recruited CD8+ T cells expressed granzyme B (GzmB). Furthermore, gzmB-/- mice harbored reduced parasite numbers in the brain as a consequence of enhanced antiparasitic CD4+ T cell responses and were protected from ECM. We showed in these ECM-resistant mice that adoptively transferred, Ag-specific CD8+ T cells migrated to the brain, but did not induce ECM until a critical Ag threshold was reached. ECM induction was exquisitely dependent on Ag-specific CD8+ T cell-derived perforin and GzmB, but not IFN-γ. In wild-type mice, full activation of brain-recruited CD8+ T cells also depended on a critical number of parasites in this tissue, which in turn, was sustained by these tissue-recruited cells. Thus, an interdependent relationship between parasite burden and CD8+ T cells dictates the onset of perforin/GzmB-mediated ECM. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
14. Elucidating the Motif for CpG Oligonucleotide Binding to the Dendritic Cell Receptor DEC-205 Leads to Improved Adjuvants for Liver-Resident Memory.
- Author
-
Li J, Panetta F, O'Keeffe M, Leal Rojas IM, Radford KJ, Zhang JG, Fernandez-Ruiz D, Davey GM, Gully BS, Tullett KM, Rossjohn J, Berry R, Lee CN, Lahoud MH, Heath WR, and Caminschi I
- Subjects
- Animals, Dendritic Cells, Interleukin-12, Liver, Mice, Adjuvants, Immunologic, Oligodeoxyribonucleotides
- Abstract
DEC-205 is a cell-surface receptor that transports bound ligands into the endocytic pathway for degradation or release within lysosomal endosomes. This receptor has been reported to bind a number of ligands, including keratin, and some classes of CpG oligodeoxynucleotides (ODN). In this study, we explore in detail the requirements for binding ODNs, revealing that DEC-205 efficiently binds single-stranded, phosphorothioated ODN of ≥14 bases, with preference for the DNA base thymidine, but with no requirement for a CpG motif. DEC-205 fails to bind double-stranded phosphodiester ODN, and thus does not bind the natural type of DNA found in mammals. The ODN binding preferences of DEC-205 result in strong binding of B class ODN, moderate binding to C class ODN, minimal binding to P class ODN, and no binding to A class ODN. Consistent with DEC-205 binding capacity, induction of serum IL-12p70 or activation of B cells by each class of ODN correlated with DEC-205 dependence in mice. Thus, the greater the DEC-205 binding capacity, the greater the dependence on DEC-205 for optimal responses. Finally, by covalently linking a B class ODN that efficiently binds DEC-205, to a P class ODN that shows poor binding, we improved DEC-205 binding and increased adjuvancy of the hybrid ODN. The hybrid ODN efficiently enhanced induction of effector CD8 T cells in a DEC-205-dependent manner. Furthermore, the hybrid ODN induced robust memory responses, and was particularly effective at promoting the development of liver tissue-resident memory T cells., (Copyright © 2021 by The American Association of Immunologists, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
15. CD8 + and CD4 + T Cells Infiltrate into the Brain during Plasmodium berghei ANKA Infection and Form Long-Term Resident Memory.
- Author
-
Ghazanfari N, Gregory JL, Devi S, Fernandez-Ruiz D, Beattie L, Mueller SN, and Heath WR
- Subjects
- Adoptive Transfer methods, Animals, Disease Models, Animal, Female, Malaria, Cerebral parasitology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Parasitemia immunology, Spleen immunology, Blood-Brain Barrier immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Malaria, Cerebral immunology, Plasmodium berghei immunology
- Abstract
In the Plasmodium berghei ANKA mouse model of malaria, accumulation of CD8
+ T cells and infected RBCs in the brain promotes the development of experimental cerebral malaria (ECM). In this study, we used malaria-specific transgenic CD4+ and CD8+ T cells to track evolution of T cell immunity during the acute and memory phases of P. berghei ANKA infection. Using a combination of techniques, including intravital multiphoton and confocal microscopy and flow cytometric analysis, we showed that, shortly before onset of ECM, both CD4+ and CD8+ T cell populations exit the spleen and begin infiltrating the brain blood vessels. Although dominated by CD8+ T cells, a proportion of both T cell subsets enter the brain parenchyma, where they are largely associated with blood vessels. Intravital imaging shows these cells moving freely within the brain parenchyma. Near the onset of ECM, leakage of RBCs into areas of the brain can be seen, implicating severe damage. If mice are cured before ECM onset, brain infiltration by T cells still occurs, but ECM is prevented, allowing development of long-term resident memory T cell populations within the brain. This study shows that infiltration of malaria-specific T cells into the brain parenchyma is associated with cerebral immunopathology and the formation of brain-resident memory T cells. The consequences of these resident memory populations is unclear but raises concerns about pathology upon secondary infection., (Copyright © 2021 by The American Association of Immunologists, Inc.)- Published
- 2021
- Full Text
- View/download PDF
16. Display of Native Antigen on cDC1 That Have Spatial Access to Both T and B Cells Underlies Efficient Humoral Vaccination.
- Author
-
Kato Y, Steiner TM, Park HY, Hitchcock RO, Zaid A, Hor JL, Devi S, Davey GM, Vremec D, Tullett KM, Tan PS, Ahmet F, Mueller SN, Alonso S, Tarlinton DM, Ploegh HL, Kaisho T, Beattie L, Manton JH, Fernandez-Ruiz D, Shortman K, Lahoud MH, Heath WR, and Caminschi I
- Subjects
- Animals, Antigen Presentation, Autoantigens immunology, Autoantigens metabolism, Cell Differentiation, Cells, Cultured, Cytokines metabolism, Immunity, Humoral, Lectins, C-Type genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, Immunologic genetics, Vaccination, B-Lymphocytes immunology, Dendritic Cells immunology, Lectins, C-Type metabolism, Receptors, Immunologic metabolism, Th1 Cells immunology, Th2 Cells immunology
- Abstract
Follicular dendritic cells and macrophages have been strongly implicated in presentation of native Ag to B cells. This property has also occasionally been attributed to conventional dendritic cells (cDC) but is generally masked by their essential role in T cell priming. cDC can be divided into two main subsets, cDC1 and cDC2, with recent evidence suggesting that cDC2 are primarily responsible for initiating B cell and T follicular helper responses. This conclusion is, however, at odds with evidence that targeting Ag to Clec9A (DNGR1), expressed by cDC1, induces strong humoral responses. In this study, we reveal that murine cDC1 interact extensively with B cells at the border of B cell follicles and, when Ag is targeted to Clec9A, can display native Ag for B cell activation. This leads to efficient induction of humoral immunity. Our findings indicate that surface display of native Ag on cDC with access to both T and B cells is key to efficient humoral vaccination., (Copyright © 2020 by The American Association of Immunologists, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
17. Development of a Novel CD4 + TCR Transgenic Line That Reveals a Dominant Role for CD8 + Dendritic Cells and CD40 Signaling in the Generation of Helper and CTL Responses to Blood-Stage Malaria.
- Author
-
Fernandez-Ruiz D, Lau LS, Ghazanfari N, Jones CM, Ng WY, Davey GM, Berthold D, Holz L, Kato Y, Enders MH, Bayarsaikhan G, Hendriks SH, Lansink LIM, Engel JA, Soon MSF, James KR, Cozijnsen A, Mollard V, Uboldi AD, Tonkin CJ, de Koning-Ward TF, Gilson PR, Kaisho T, Haque A, Crabb BS, Carbone FR, McFadden GI, and Heath WR
- Subjects
- Animals, Antigens, Protozoan immunology, CD40 Antigens deficiency, CD40 Ligand immunology, Cells, Cultured, Crosses, Genetic, Hybridomas, Lymphocyte Activation, Malaria, Cerebral immunology, Malaria, Cerebral prevention & control, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic genetics, Plasmodium berghei immunology, Radiation Chimera, Antigen Presentation, CD4-Positive T-Lymphocytes immunology, CD40 Antigens immunology, Dendritic Cells immunology, Malaria immunology, Mice, Transgenic immunology, Parasitemia immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
We describe an MHC class II (I-A
b )-restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human ( Plasmodium falciparum ) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC., (Copyright © 2017 by The American Association of Immunologists, Inc.)- Published
- 2017
- Full Text
- View/download PDF
18. Pillars article: T cell receptor antagonist peptides induce positive selection. Cell. 1994. 76: 17-27.
- Author
-
Hogquist KA, Jameson SC, Heath WR, Howard JL, Bevan MJ, and Carbone FR
- Subjects
- Animals, Cell Differentiation genetics, Cell Differentiation immunology, History, 20th Century, Mice, Mice, Knockout, Mice, Transgenic, Organ Culture Techniques, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets metabolism, Thymus Gland embryology, Thymus Gland immunology, beta 2-Microglobulin deficiency, beta 2-Microglobulin genetics, Peptide Fragments physiology, Receptors, Antigen, T-Cell antagonists & inhibitors, Receptors, Antigen, T-Cell physiology, T-Lymphocyte Subsets immunology, Thymus Gland cytology
- Published
- 2012
19. Cross-priming: its beginnings.
- Author
-
Carbone FR and Heath WR
- Subjects
- Animals, H-2 Antigens immunology, History, 20th Century, History, 21st Century, Mice, Mice, Inbred BALB C immunology, Mice, Inbred C57BL immunology, Minor Histocompatibility Antigens history, Minor Histocompatibility Antigens immunology, Peptides immunology, T-Lymphocytes, Cytotoxic immunology, Antigen Presentation immunology, Cross-Priming immunology, H-2 Antigens history, Peptides history
- Published
- 2010
- Full Text
- View/download PDF
20. Cutting edge: priming of CD8 T cell immunity to herpes simplex virus type 1 requires cognate TLR3 expression in vivo.
- Author
-
Davey GM, Wojtasiak M, Proietto AI, Carbone FR, Heath WR, and Bedoui S
- Subjects
- Adaptor Proteins, Vesicular Transport genetics, Adaptor Proteins, Vesicular Transport immunology, Adaptor Proteins, Vesicular Transport metabolism, Animals, Bone Marrow Cells cytology, Bone Marrow Cells immunology, Bone Marrow Cells metabolism, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Dendritic Cells cytology, Dendritic Cells immunology, Dendritic Cells metabolism, Female, Flow Cytometry, H-2 Antigens genetics, H-2 Antigens immunology, H-2 Antigens metabolism, Herpes Simplex virology, Herpesvirus 1, Human growth & development, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Inbred Strains, Mice, Knockout, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 immunology, Myeloid Differentiation Factor 88 metabolism, Reverse Transcriptase Polymerase Chain Reaction, T-Lymphocytes, Cytotoxic cytology, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Cytotoxic metabolism, Toll-Like Receptor 3 genetics, Toll-Like Receptor 3 metabolism, CD8-Positive T-Lymphocytes immunology, Herpes Simplex immunology, Herpesvirus 1, Human immunology, Toll-Like Receptor 3 immunology
- Abstract
Despite its potential for involvement in viral immunity, little evidence links TLR3 to adaptive antiviral responses. Here we show that TLR3 is required for the generation of CD8 T cell immunity to HSV-1. The magnitude of the gB-specific CD8 T cell response after flank infection by HSV-1 was significantly reduced in mice lacking TIR domain-containing adaptor-inducing IFN-beta or TLR3, but not MyD88. Impaired CTL induction was evident in chimeric mice lacking TLR3 in bone marrow (BM)-derived cells. Among the dendritic cell subsets, TLR3 was expressed by CD8alpha(+) dendritic cells, known to be involved in priming HSV-1-specific CD8 T cells. Use of mixed BM chimeras revealed that TLR3 and the MHC class I-restriction element must be expressed by the same BM-derived cell for effective priming. These data imply that a cognate linkage between TLR3 and MHC class I is required for efficient CTL priming to HSV-1.
- Published
- 2010
- Full Text
- View/download PDF
21. The C-type lectin Clec12A present on mouse and human dendritic cells can serve as a target for antigen delivery and enhancement of antibody responses.
- Author
-
Lahoud MH, Proietto AI, Ahmet F, Kitsoulis S, Eidsmo L, Wu L, Sathe P, Pietersz S, Chang HW, Walker ID, Maraskovsky E, Braley H, Lew AM, Wright MD, Heath WR, Shortman K, and Caminschi I
- Subjects
- Animals, Antigen Presentation immunology, Cell Membrane immunology, Cells, Cultured, Humans, Leukocytes immunology, Mice, Antibody Formation immunology, Antigens immunology, Dendritic Cells immunology, Lectins, C-Type immunology, Receptors, Mitogen immunology
- Abstract
We have cloned the mouse and human C-type lectin Clec12A, expressed both, and produced mAb recognizing both. Mouse Clec12A is highly expressed on splenic CD8(+) dendritic cells (DC) and plasmacytoid DC. A proportion of CD8(-)DC also expresses lower levels of Clec12A, as do monocytes, macrophages, and B cells. Human CLEC12A, like the mouse counterpart, is expressed on blood monocytes and DC, including pDC and BDCA-3(+)DC, the proposed equivalent of mouse CD8(+)DC. To determine whether Ag targeted to Clec12A could induce immune responses, mice were injected with a rat mAb recognizing Clec12A, or a control rat mAb, then production of anti-rat Ig was measured. Anti-Clec12A mAb alone produced only moderate responses, but these were amplified by coinjecting only small amounts of LPS as a DC activation agent. Furthermore, when OVA was conjugated to anti-Clec12A mAb, OVA-specific T cells were induced to proliferate. This Ag presentation to naive T cells was due to targeting conventional DC, because their ablation eliminated T cell activation. The potent Ab responses induced using microgram amounts of anti-Clec12A and minimal amounts of adjuvant demonstrate that this molecule can be used as an Ag-delivery target to enhance Ab responses to vaccines.
- Published
- 2009
- Full Text
- View/download PDF
22. Characterization of an immediate splenic precursor of CD8+ dendritic cells capable of inducing antiviral T cell responses.
- Author
-
Bedoui S, Prato S, Mintern J, Gebhardt T, Zhan Y, Lew AM, Heath WR, Villadangos JA, and Segura E
- Subjects
- Adoptive Transfer, Animals, Antigen Presentation immunology, Blotting, Western, CD24 Antigen immunology, CD24 Antigen metabolism, CD8 Antigens immunology, CD8 Antigens metabolism, Cell Differentiation, Dendritic Cells cytology, Dendritic Cells metabolism, Flow Cytometry, Histocompatibility Antigens Class I, Mice, Mice, Transgenic, Spleen cytology, Spleen immunology, Stem Cells cytology, Stem Cells metabolism, T-Lymphocytes virology, Cross-Priming immunology, Dendritic Cells immunology, Herpesvirus 1, Human immunology, Lymphocyte Activation immunology, Stem Cells immunology, T-Lymphocytes immunology
- Abstract
Mouse spleens contain three major dendritic cell (DC) populations: plasmacytoid DC, conventional CD8(+)CD24(+) DC (CD8(+) DC), and conventional CD8(-)CD24(-) DC (CD8(-) DC). We have previously shown that CD8(+) DC are the major cross-presenting subtype in vivo and are the main inducers of antiviral cytotoxic T lymphocyte responses. Here we show that after depletion of CD8(+) DC, the only DC capable of viral Ag presentation was a small subset that expresses CD24 but not CD8. This CD8(-)CD24(+) DC population is greatly expanded in mice treated with the DC growth factor FMS-like tyrosine kinase 3 ligand. The CD8(-)CD24(+) DC represent an immediate precursor of CD8(+) DC, as demonstrated by their expression pattern of characteristic markers of CD8(+) DC, their capacity to cross-present in vitro, and their conversion into CD8(+) DC upon adoptive transfer into recipient mice. Accordingly, the lifespan of transferred CD8(-)CD24(+) DC in vivo was greatly enhanced as compared with terminally differentiated CD8(+) DC. Moreover, in a vaccination protocol, CD8(-)CD24(+) DC induced stronger T cell responses and accelerated viral clearance of HSV-1 compared with CD8(+) DC. Our results demonstrate that the ability to cross-present first appears in an immediate precursor population of CD8(+) DC that does not yet express CD8. The enhanced capacity of CD8(-)CD24(+) DC to induce immune responses upon adoptive transfer makes them an attractive novel tool for DC-based immunotherapies.
- Published
- 2009
- Full Text
- View/download PDF
23. Aire-deficient C57BL/6 mice mimicking the common human 13-base pair deletion mutation present with only a mild autoimmune phenotype.
- Author
-
Hubert FX, Kinkel SA, Crewther PE, Cannon PZ, Webster KE, Link M, Uibo R, O'Bryan MK, Meager A, Forehan SP, Smyth GK, Mittaz L, Antonarakis SE, Peterson P, Heath WR, and Scott HS
- Subjects
- Amino Acid Sequence, Animals, Base Pairing genetics, Base Sequence, Cell Line, Disease Models, Animal, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Knockout, Molecular Sequence Data, Polyendocrinopathies, Autoimmune immunology, Polyendocrinopathies, Autoimmune metabolism, Sequence Homology, Amino Acid, Thymus Gland immunology, Thymus Gland metabolism, Thymus Gland pathology, Transcription Factors biosynthesis, AIRE Protein, Molecular Mimicry genetics, Molecular Mimicry immunology, Mutagenesis, Site-Directed, Phenotype, Polyendocrinopathies, Autoimmune genetics, Transcription Factors deficiency, Transcription Factors genetics
- Abstract
Autoimmune regulator (AIRE) is an important transcription regulator that mediates a role in central tolerance via promoting the "promiscuous" expression of tissue-specific Ags in the thymus. Although several mouse models of Aire deficiency have been described, none has analyzed the phenotype induced by a mutation that emulates the common 13-bp deletion in human APECED (autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy) by disrupting the first plant homeodomain in exon 8. Aire-deficient mice with a corresponding mutation showed some disturbance of the medullary epithelial compartment, but at the phenotypic level their T cell compartment appeared relatively normal in the thymus and periphery. An increase in the number of activated T cells was evident, and autoantibodies against several organs were detected. At the histological level, lymphocytic infiltration of several organs indicated the development of autoimmunity, although symptoms were mild and the quality of life for Aire-deficient mice appeared equivalent to wild-type littermates, with the exception of male infertility. Vbeta and CDR3 length analysis suggested that each Aire-deficient mouse developed its own polyclonal autoimmune repertoire. Finally, given the prevalence of candidiasis in APECED patients, we examined the control of infection with Candida albicans in Aire-deficient mice. No increase in disease susceptibility was found for either oral or systemic infection. These observations support the view that additional genetic and/or environmental factors contribute substantially to the overt nature of autoimmunity associated with Aire mutations, even for mutations identical to those found in humans with APECED.
- Published
- 2009
- Full Text
- View/download PDF
24. Differential migration of epidermal and dermal dendritic cells during skin infection.
- Author
-
Eidsmo L, Allan R, Caminschi I, van Rooijen N, Heath WR, and Carbone FR
- Subjects
- Animals, Cell Aggregation immunology, Dendritic Cells pathology, Dendritic Cells virology, Dermis pathology, Dermis virology, Epidermis pathology, Epidermis virology, Langerhans Cells immunology, Langerhans Cells pathology, Langerhans Cells virology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Monocytes immunology, Monocytes pathology, Monocytes virology, Stem Cells immunology, Stem Cells pathology, Stem Cells virology, Dendritic Cells immunology, Dermis immunology, Epidermis immunology, Herpes Simplex immunology, Herpes Simplex pathology, Herpesvirus 1, Human immunology
- Abstract
Dendritic cells (DCs) are extremely heterogeneous, most evident in the skin where a variety of different subsets have been identified in recent years. DCs of healthy skin include a number of distinct populations in the dermal layer as well as the well-characterized Langerhans cells (LCs) of the epidermis. These steady-state populations are augmented during bouts of local inflammation by additional monocyte-derived DCs. In an effort to better understand the distinction between the different subsets, we examined their behavior following skin infection with HSV. LC emigration rapidly followed appearance of virus in the skin and resulted in depopulation of regions in areas surrounding infected nerve endings. A separate DC population was found to accumulate within the dermis under patches of active epidermal infection with at least some derived from blood monocyte precursors. Ag-positive DCs could occasionally be found in these dermal accumulations, although they represented a minority of DCs in these areas. In addition, infected DCs appeared compromised in their trafficking capabilities and were largely absent from the migrating population. On resolution of skin disease, LCs repopulated the reformed epidermis and these were of mixed origin, with around half entering from the circulation and the remainder derived from local progenitors. Overall, our results show a range of migrational complexities between distinct skin DC populations as a consequence of localized infection.
- Published
- 2009
- Full Text
- View/download PDF
25. Cutting edge: local recall responses by memory T cells newly recruited to peripheral nonlymphoid tissues.
- Author
-
Wakim LM, Gebhardt T, Heath WR, and Carbone FR
- Subjects
- Animals, Cell Aggregation immunology, Cell Differentiation immunology, Cell Proliferation, Epitopes, T-Lymphocyte metabolism, Ganglia, Spinal cytology, Ganglia, Spinal immunology, Ganglia, Spinal virology, Herpes Simplex immunology, Herpes Simplex prevention & control, Herpes Simplex virology, Herpesvirus 1, Human immunology, Lymph Nodes cytology, Lymph Nodes immunology, Lymph Nodes transplantation, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, T-Lymphocyte Subsets cytology, Cell Movement immunology, Immunologic Memory, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets virology
- Abstract
Infection results in the formation of a circulating effector memory T cell population able to enter peripheral tissues either in the steady state or in response to localized infection. As a consequence, recall is thought to result from a phased response first involving those T cells already at the site of infection followed by the infiltration of memory cells from the wider circulation. We have recently reported that tissue-resident T cells can undergo stimulation and proliferation in response to local infection. In this study, we examine the proliferation of memory T cells newly recruited from the circulation. Our results show that although recruitment of circulating memory cells is nonspecific in nature, there is preferential proliferation of specific T cells within infected tissues. Thus, expansion represents a means of local Ag-specific enrichment of T cells recruited from a circulating memory pool of mixed specificities.
- Published
- 2008
- Full Text
- View/download PDF
26. Cutting edge: Enhanced IL-2 signaling can convert self-specific T cell response from tolerance to autoimmunity.
- Author
-
Waithman J, Gebhardt T, Davey GM, Heath WR, and Carbone FR
- Subjects
- Animals, Antigen Presentation physiology, Cell Movement immunology, Dendritic Cells cytology, Dendritic Cells immunology, Granzymes immunology, Immunologic Memory physiology, Mice, Receptors, Interleukin-2 immunology, Signal Transduction, Skin cytology, T-Lymphocytes cytology, Autoantigens immunology, Autoimmunity physiology, Immune Tolerance physiology, Interleukin-2 immunology, Skin immunology, T-Lymphocytes immunology
- Abstract
Naive and memory T cells show differences in their response to antigenic stimulation. We examined whether this difference extended to the peripheral deletion of T cells reactive to self-Ag or, alternatively, the induction of autoimmunity. Our results show that although both populations where susceptible to deletion, memory T cells, but not naive T cells, also gave rise to autoimmunity after in vivo presentation of skin-derived self-Ags. The same migratory dendritic cells presented self-Ag to both naive and memory T cell populations, but only the latter had significant levels of the effector molecule granzyme B. Memory T cells also expressed increased levels of the high affinity IL-2 receptor chain after self-Ag recognition. Provision of IL-2 signaling using a stimulatory complex of anti-IL-2 Ab and IL-2 drove the otherwise tolerant naive T cells toward an autoimmune response. Therefore, enhanced IL-2 signaling can act as a major selector between tolerance and autoimmunity.
- Published
- 2008
- Full Text
- View/download PDF
27. A specific anti-Aire antibody reveals aire expression is restricted to medullary thymic epithelial cells and not expressed in periphery.
- Author
-
Hubert FX, Kinkel SA, Webster KE, Cannon P, Crewther PE, Proeitto AI, Wu L, Heath WR, and Scott HS
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal biosynthesis, Antigen-Antibody Reactions, Cell Nucleus immunology, Cell Nucleus metabolism, Immunophenotyping, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Molecular Sequence Data, Quantum Dots, Rats, Thymus Gland metabolism, Transcription Factors deficiency, Transcription Factors genetics, AIRE Protein, Antibodies, Monoclonal analysis, Antibody Specificity, Epithelial Cells immunology, Epithelial Cells metabolism, Thymus Gland cytology, Thymus Gland immunology, Transcription Factors biosynthesis, Transcription Factors immunology
- Abstract
Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy is an autoimmune disorder caused by mutations in the autoimmune regulator gene AIRE. We examined the expression of Aire in different organs (thymus, spleen, and lymph nodes) in C57BL/6 mice, using a novel rat mAb, specific for murine Aire. Using flow cytometry, directly fluorochrome-labeled mAb revealed Aire expression in a rare thymic cellular subset that was CD45(-), expressed low levels of Ly51, and was high for MHC-II and EpCam. This subset also expressed a specific pattern of costimulatory molecules, including CD40, CD80, and PD-L1. Immunohistochemical analysis revealed that Aire(+) cells were specifically localized to the thymus or, more precisely, to the cortico-medulla junction and medulla, correlating with the site of negative selection. Although in agreement with previous studies, low levels of Aire mRNA was detected in all dendritic cell subtypes however lacZ staining, immunohistochemistry and flow cytometry failed to detect Aire protein. At a cellular level, Aire was expressed in perinuclear speckles within the nucleus. This report provides the first detailed analysis of Aire protein expression, highlighting the precise location at both the tissue and cellular level.
- Published
- 2008
- Full Text
- View/download PDF
28. Skin-derived dendritic cells can mediate deletional tolerance of class I-restricted self-reactive T cells.
- Author
-
Waithman J, Allan RS, Kosaka H, Azukizawa H, Shortman K, Lutz MB, Heath WR, Carbone FR, and Belz GT
- Subjects
- Animals, Antigen-Presenting Cells immunology, Antigens immunology, Cell Differentiation, Cell Proliferation, Cells, Cultured, Coculture Techniques, Langerhans Cells immunology, Mice, Mice, Transgenic, Ovalbumin immunology, T-Lymphocytes cytology, Autoimmunity immunology, Dendritic Cells cytology, Dendritic Cells immunology, Histocompatibility Antigens Class I immunology, Immune Tolerance immunology, Skin immunology, T-Lymphocytes immunology
- Abstract
Skin-draining lymph nodes contain a number of dendritic cell (DC) subsets of different origins. Some of these are migratory, such as the skin-derived epidermal Langerhans cells and a separate dermal DC subset, whereas others are lymphoid resident in nature, such as the CD8+ DCs found throughout the lymphoid tissues. In this study, we examine the DC subset presentation of skin-derived self-Ag by migratory and lymphoid-resident DCs, both in the steady state and under conditions of local skin infection. We show that presentation of self-Ag is confined to skin-derived migrating DCs in both settings. Steady state presentation resulted in deletional T cell tolerance despite these DCs expressing a relatively mature phenotype as measured by traditional markers such as the level of MHC class II and CD86 expression. Thus, self-Ag can be carried to the draining lymph nodes by skin-derived DCs and there presented by these same cells for tolerization of the circulating T cell pool.
- Published
- 2007
- Full Text
- View/download PDF
29. Cutting edge: central memory T cells do not show accelerated proliferation or tissue infiltration in response to localized herpes simplex virus-1 infection.
- Author
-
Stock AT, Jones CM, Heath WR, and Carbone FR
- Subjects
- Animals, Herpes Simplex immunology, Herpes Simplex pathology, Kinetics, Lymph Nodes immunology, Lymph Nodes pathology, Lymph Nodes virology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Resting Phase, Cell Cycle immunology, Skin Diseases, Viral immunology, Skin Diseases, Viral pathology, T-Lymphocyte Subsets pathology, Cell Movement immunology, Cell Proliferation, Herpesvirus 1, Human immunology, Immunologic Memory, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets virology
- Abstract
Memory T cells mount an enhanced response to secondary infections. Such an enhancement has been attributed in part to the ability of memory cells to more rapidly respond to cognate stimulation. In this study we have examined the rapidity with which murine CD8(+) memory T cells respond to a localized infection with HSV. Although central memory T cells (TcM), but not the effector memory T cells, mounted a strong recall response to secondary infection, the kinetics of TcM proliferation, the magnitude of their expansion, and their infiltration into infected nonlymphoid tissues were not advanced compared with that observed for naive T cells. These findings imply that it is the lack of accelerated proliferation kinetics and the subsequent delayed dissemination into the periphery that limits the ability of TcM to rapidly control localized virus replication.
- Published
- 2006
- Full Text
- View/download PDF
30. CD4+ T cells can protect APC from CTL-mediated elimination.
- Author
-
Mueller SN, Jones CM, Stock AT, Suter M, Heath WR, and Carbone FR
- Subjects
- Adoptive Transfer, Amino Acid Sequence, Animals, Antigen Presentation immunology, Antigen-Presenting Cells metabolism, Antigen-Presenting Cells virology, CD4-Positive T-Lymphocytes transplantation, CD4-Positive T-Lymphocytes virology, Cell Differentiation immunology, Cells, Cultured, Dendritic Cells cytology, Dendritic Cells immunology, Dendritic Cells metabolism, Dendritic Cells virology, Herpesvirus 8, Human immunology, Lymph Nodes immunology, Lymph Nodes metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Molecular Sequence Data, T-Lymphocytes, Cytotoxic transplantation, Viral Envelope Proteins immunology, Viral Envelope Proteins metabolism, Antigen-Presenting Cells cytology, Antigen-Presenting Cells immunology, CD4-Positive T-Lymphocytes immunology, Cytotoxicity, Immunologic, Lymphocyte Depletion, T-Lymphocytes, Cytotoxic immunology
- Abstract
Professional APC play a central role in generating antiviral CD8(+) CTL immunity. However, the fate of such APC following interaction with these same CTL remains poorly understood. We have shown previously that prolonged Ag presentation persists in the presence of a strong CTL response following HSV infection. In this study, we examined the mechanism of survival of APC in vivo when presenting an immunodominant determinant from HSV. We show that transferred peptide-labeled dendritic cells were eliminated from draining lymph nodes in the presence of HSV-specific CTL. Maturation of dendritic cells with LPS or anti-CD40 before injection protected against CTL lysis in vivo. Furthermore, endogenous APC could be eliminated from draining lymph nodes early after HSV infection by adoptive transfer of HSV-specific CTL, yet the cotransfer of significant virus-specific CD4(+) T cell help promoted prolonged Ag presentation. This suggests that Th cells may assist in prolonging class I-restricted Ag presentation, potentially enhancing CTL recruitment and allowing more efficient T cell priming.
- Published
- 2006
- Full Text
- View/download PDF
31. CD8alpha+ dendritic cells selectively present MHC class I-restricted noncytolytic viral and intracellular bacterial antigens in vivo.
- Author
-
Belz GT, Shortman K, Bevan MJ, and Heath WR
- Subjects
- Animals, Antigens, Bacterial metabolism, Antigens, Viral metabolism, CD8-Positive T-Lymphocytes immunology, Histocompatibility Antigens Class I metabolism, Listeria monocytogenes immunology, Lymphocytic choriomeningitis virus immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Antigen Presentation, CD8 Antigens metabolism, Dendritic Cells classification, Dendritic Cells immunology
- Abstract
CD8alpha(+) dendritic cells (DCs) have been shown to be the principal DC subset involved in priming MHC class I-restricted CTL immunity to a variety of cytolytic viruses, including HSV type 1, influenza, and vaccinia virus. Whether priming of CTLs by CD8alpha(+) DCs is limited to cytolytic viruses, which may provide dead cellular material for this DC subset, or whether these DCs selectively present intracellular Ags, is unknown. To address this question, we examined Ag presentation to a noncytolytic virus, lymphocytic choriomeningitis virus, and to an intracellular bacterium, Listeria monocytogenes. We show that regardless of the type of intracellular infection, CD8alpha(+) DCs are the principal DC subset that initiate CD8(+) T cell immunity.
- Published
- 2005
- Full Text
- View/download PDF
32. Cutting edge: prolonged antigen presentation after herpes simplex virus-1 skin infection.
- Author
-
Stock AT, Mueller SN, van Lint AL, Heath WR, and Carbone FR
- Subjects
- Animals, CD8-Positive T-Lymphocytes, Flow Cytometry, Mice, Time Factors, Antigen Presentation immunology, Cytotoxicity, Immunologic, Herpes Simplex immunology, Herpesvirus 1, Human immunology, Lymphocyte Activation immunology
- Abstract
It has been reported that MHC class I-restricted Ag presentation persists for only a short period following infection with certain pathogens, declining in parallel with the emergence of specific CTL activity. We have examined this issue in the case of murine infection with HSV-1. We found that the period of Ag presentation capable of priming naive CD8(+) T cells is comparatively prolonged, persisting for at least 7 days after infection, and continuing despite the appearance of localized CTL activity. Ag presentation was abbreviated to 3 or 4 days postinfection by surgical excision of the inoculation site early after infection. This intervention attenuated the size of the primary CTL response, implying that prolonged presentation is necessary to drive maximal CTL expansion. Combined, these data show that, in some types of infection, CTL priming can extend well beyond the first 24-48 h after primary inoculation.
- Published
- 2004
- Full Text
- View/download PDF
33. Helper requirements for generation of effector CTL to islet beta cell antigens.
- Author
-
Behrens GM, Li M, Davey GM, Allison J, Flavell RA, Carbone FR, and Heath WR
- Subjects
- Adoptive Transfer, Animals, Antigen Presentation genetics, CD40 Ligand genetics, CD40 Ligand physiology, Cell Communication genetics, Cell Division genetics, Cell Division immunology, Cells, Cultured, Diabetes Mellitus, Experimental genetics, Diabetes Mellitus, Experimental immunology, Diabetes Mellitus, Experimental pathology, Epitopes, T-Lymphocyte genetics, Epitopes, T-Lymphocyte immunology, Epitopes, T-Lymphocyte metabolism, Histocompatibility Antigens Class II genetics, Histocompatibility Antigens Class II immunology, Histocompatibility Antigens Class II metabolism, Lymphocyte Activation genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Ovalbumin immunology, Ovalbumin metabolism, T-Lymphocytes, Cytotoxic transplantation, T-Lymphocytes, Helper-Inducer metabolism, Autoantigens physiology, Cell Communication immunology, Cytotoxicity, Immunologic genetics, Islets of Langerhans immunology, Islets of Langerhans pathology, Lymphocyte Activation immunology, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Helper-Inducer immunology
- Abstract
We have dissected the helper requirements for converting a tolerogenic CD8 T cell response into one capable of causing destruction of the pancreatic islets. Injection of naive OVA-specific CD8 T cells into transgenic mice expressing OVA in the pancreas only resulted in islet destruction when activated CD4 Th cells were coinjected. This requirement for activated CD4 T cell help for induction of primary CD8 T cell-mediated immunity to tissue Ags contrasts recent reports suggesting that help is only important for CTL memory. Our findings show that signaling of CD40 on the dendritic cell presenting to CD8 T cells is important, but not sufficient, for induction of diabetes. Furthermore, once helpers are activated, they need not recognize Ag on the dendritic cells they license. This provides insight into the helper requirements for adoptive transfer immunotherapy of tumors and suggests key points for inhibition of CTL-mediated autoimmunity.
- Published
- 2004
- Full Text
- View/download PDF
34. Cutting edge: conventional CD8 alpha+ dendritic cells are generally involved in priming CTL immunity to viruses.
- Author
-
Belz GT, Smith CM, Eichner D, Shortman K, Karupiah G, Carbone FR, and Heath WR
- Subjects
- Animals, Cell Division immunology, Cell Separation, Dendritic Cells metabolism, Immunity, Cellular, Injections, Intravenous, Injections, Subcutaneous, Interphase immunology, Lymph Nodes cytology, Lymph Nodes immunology, Lymph Nodes virology, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Spleen cytology, Spleen immunology, Spleen virology, CD8 Antigens biosynthesis, Dendritic Cells immunology, Dendritic Cells virology, Herpes Simplex immunology, Orthomyxoviridae Infections immunology, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Cytotoxic virology, Vaccinia immunology
- Abstract
Dendritic cells (DCs) play a central role in initiating immune responses. Despite this, there is little understanding how different DC subsets contribute to immunity to different pathogens. CD8alpha(+) DC have been shown to prime immunity to HSV. Whether this very limited capacity of a single DC subset priming CTL immunity is restricted to HSV infection or is a more general property of anti-viral immunity was examined. Here, we show that the CD8alpha(+) DCs are the principal DC subset that initiates CTL immunity to s.c. infection by influenza virus, HSV, and vaccinia virus. This same subset also dominated immunity after i.v. infection with all three viruses, suggesting a similar involvement in other routes of infection. These data highlight the general role played by CD8alpha(+) DCs in CTL priming to viral infection and raises the possibility that this DC subset is specialized for viral immunity.
- Published
- 2004
- Full Text
- View/download PDF
35. Herpes simplex virus-specific CD8+ T cells can clear established lytic infections from skin and nerves and can partially limit the early spread of virus after cutaneous inoculation.
- Author
-
van Lint A, Ayers M, Brooks AG, Coles RM, Heath WR, and Carbone FR
- Subjects
- Administration, Cutaneous, Adoptive Transfer, Animals, CD8-Positive T-Lymphocytes transplantation, Disease Progression, Epitopes, T-Lymphocyte immunology, Female, Ganglia, Spinal pathology, Ganglia, Spinal virology, Herpes Simplex immunology, Herpes Simplex pathology, Herpesvirus 1, Human growth & development, Hindlimb, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Peripheral Nervous System Diseases immunology, Peripheral Nervous System Diseases pathology, Receptors, Antigen, T-Cell genetics, Skin Diseases, Viral immunology, Skin Diseases, Viral pathology, Virus Replication immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes virology, Ganglia, Spinal immunology, Herpes Simplex prevention & control, Herpesvirus 1, Human immunology, Peripheral Nervous System Diseases prevention & control, Skin Diseases, Viral prevention & control, Viral Envelope Proteins immunology
- Abstract
HSV infects skin or mucosal epithelium as well as entering the sensory nerves and ganglia. We have used TCR-transgenic T cells specific for the immunodominant class I-restricted determinant from HSV glycoprotein B (gB) combined with a flank zosteriform model of infection to examine the ability of CD8+ T cells to deal with infection. During the course of zosteriform disease, virus rapidly spreads from the primary inoculation site in the skin to sensory dorsal root ganglia and subsequently reappears in the distal flank. Virus begins to be cleared from all sites about 5 days after infection when gB-specific CD8+ T cells first appear within infected tissues. Although activated gB-specific effectors can partially limit virus egress from the skin, they do so only at the earliest times after infection and are ineffective at halting the progression of zosteriform disease once virus has left the inoculation site. In contrast, these same T cells can completely clear ongoing lytic replication if transferred into infected immunocompromised RAG-1-/- mice. Therefore, we propose that the role of CD8+ T cells during the normal course of disease is to clear replicating virus after infection is well established rather than limit the initial spread of HSV from the primary site of inoculation.
- Published
- 2004
- Full Text
- View/download PDF
36. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells.
- Author
-
Nowak AK, Lake RA, Marzo AL, Scott B, Heath WR, Collins EJ, Frelinger JA, and Robinson BW
- Subjects
- Animals, Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, Antigens, Neoplasm metabolism, Apoptosis drug effects, CD8-Positive T-Lymphocytes metabolism, Clonal Anergy drug effects, Clonal Deletion drug effects, Clonal Deletion immunology, Cytotoxicity, Immunologic drug effects, Deoxycytidine administration & dosage, Dose-Response Relationship, Immunologic, Epitopes, T-Lymphocyte immunology, Epitopes, T-Lymphocyte metabolism, Growth Inhibitors administration & dosage, Hemagglutinin Glycoproteins, Influenza Virus immunology, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Immunization, Injections, Intraperitoneal, Mesothelioma drug therapy, Mesothelioma prevention & control, Mice, Mice, Inbred BALB C, Mice, Transgenic, Tumor Cells, Cultured, Up-Regulation drug effects, Gemcitabine, Antigen Presentation drug effects, Antigens, Neoplasm immunology, Apoptosis immunology, CD8-Positive T-Lymphocytes immunology, Clonal Anergy immunology, Deoxycytidine analogs & derivatives, Mesothelioma immunology, Mesothelioma pathology, Up-Regulation immunology
- Abstract
Cross-presentation of cell-bound Ags from established, solid tumors to CD8 cells is efficient and likely to have a role in determining host response to tumor. A number of investigators have predicted that when tumor Ags are derived from apoptotic cells either no response, due to Ag "sequestration," or CD8 cross-tolerance would ensue. Because the crucial issue of whether this happens in vivo has never been addressed, we induced apoptosis of established hemagglutinin (HA)-transfected AB1 tumors in BALB/c mice using the apoptosis-inducing reagent gemcitabine. This shrank the tumor by approximately 80%. This induction of apoptosis increased cross-presentation of HA to CD8 cells yet neither gross deletion nor functional tolerance of HA-specific CD8 cells were observed, based on tetramer analysis, proliferation of specific CD8 T cells, and in vivo CTL activity. Interestingly, apoptosis primed the host for a strong antitumor response to a second, virus-generated HA-specific signal in that administration of an HA-expressing virus after gemcitabine administration markedly decreased tumor growth compared with viral administration without gemcitabine. Thus tumor cell apoptosis in vivo neither sequesters tumor Ags nor cross-tolerizes tumor-specific CD8 cells. This observation has fundamental consequences for the development of tumor immunotherapy protocols and for understanding T cell reactivity to tumors and the in vivo immune responses to apoptotic cells.
- Published
- 2003
- Full Text
- View/download PDF
37. Cutting edge: conventional CD8 alpha+ dendritic cells are preferentially involved in CTL priming after footpad infection with herpes simplex virus-1.
- Author
-
Smith CM, Belz GT, Wilson NS, Villadangos JA, Shortman K, Carbone FR, and Heath WR
- Subjects
- Animals, Antigen Presentation immunology, Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, Antigen-Presenting Cells virology, Dendritic Cells virology, Epitopes, T-Lymphocyte immunology, Epitopes, T-Lymphocyte metabolism, Herpes Simplex virology, Hindlimb, Histocompatibility Antigens Class I metabolism, Hybridomas, Immunity, Active, Immunodominant Epitopes immunology, Immunodominant Epitopes metabolism, Injections, Subcutaneous, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Viral Envelope Proteins immunology, Viral Envelope Proteins metabolism, CD8 Antigens biosynthesis, Cytotoxicity, Immunologic immunology, Dendritic Cells immunology, Dendritic Cells metabolism, Herpes Simplex immunology, Herpesvirus 1, Human immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
CTL play a major role in immunity to HSV type 1, but little is known about the priming process. In this study, we have examined the class I-restricted presentation of an immunodominant determinant from HSV-1 glycoprotein B after footpad infection. We have found that the only cell types capable of presenting this determinant in draining popliteal lymph nodes within the first 3 days after infection are the CD11c(+)CD8alpha(+)CD45RA(-) dendritic cells. Given that such class I-restricted presentation is essential for CTL priming, this implies that these conventional CD8alpha(+) dendritic cells are the key subset involved in CTL immunity to this virus.
- Published
- 2003
- Full Text
- View/download PDF
38. Suppressor of cytokine signaling-1 has IFN-gamma-independent actions in T cell homeostasis.
- Author
-
Cornish AL, Davey GM, Metcalf D, Purton JF, Corbin JE, Greenhalgh CJ, Darwiche R, Wu L, Nicola NA, Godfrey DI, Heath WR, Hilton DJ, Alexander WS, and Starr R
- Subjects
- Animals, CD4-CD8 Ratio, Carrier Proteins genetics, Epitopes, T-Lymphocyte immunology, Fetus, Homeostasis genetics, Immunophenotyping, Interferon-gamma deficiency, Interferon-gamma genetics, Lymphatic Diseases genetics, Lymphatic Diseases immunology, Lymphatic Diseases pathology, Lymphocyte Activation genetics, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Organ Culture Techniques, Receptors, Antigen, T-Cell metabolism, Signal Transduction genetics, Suppressor of Cytokine Signaling 1 Protein, Suppressor of Cytokine Signaling Proteins, T-Lymphocyte Subsets metabolism, Thymus Gland cytology, Thymus Gland immunology, Carrier Proteins physiology, Cytokines antagonists & inhibitors, Cytokines physiology, Homeostasis immunology, Interferon-gamma physiology, Repressor Proteins, Signal Transduction immunology, T-Lymphocyte Subsets immunology
- Abstract
Suppressor of cytokine signaling (SOCS)-1 is a member of a family of proteins that negatively regulate cytokine signaling pathways. We have previously established that SOCS-1 is a key regulator of IFN-gamma signaling and that IFN-gamma is responsible for the complex inflammatory disease that leads to the death of SOCS-1-deficient mice. In this study, we provide evidence that SOCS-1 is also a critical regulator of IFN-gamma-independent immunoregulatory factors. Mice lacking both SOCS-1 and IFN-gamma, although outwardly healthy, have clear abnormalities in their immune system, including a reduced ratio of CD4:CD8 T cells in lymphoid tissues and increased expression of T cell activation markers. To examine the contribution of TCR Ag specificity to these immune defects, we have generated two lines of SOCS-1-deficient mice expressing a transgenic TCR specific for an exogenous Ag, OVA (OT-I and OT-II). Although TCR transgenic SOCS-1(-/-) mice have a longer lifespan than nontransgenic SOCS-1(-/-) mice, they still die as young adults with inflammatory disease and the TCR transgenic SOCS-1(-/-) T cells appear activated despite the absence of OVA. This suggests that both Ag-dependent and -independent mechanisms contribute to the disease in SOCS-1-deficient mice. Thus, SOCS-1 is a critical regulator of T cell activation and homeostasis, and its influence extends beyond regulating IFN-gamma signaling.
- Published
- 2003
- Full Text
- View/download PDF
39. CD36 is differentially expressed by CD8+ splenic dendritic cells but is not required for cross-presentation in vivo.
- Author
-
Belz GT, Vremec D, Febbraio M, Corcoran L, Shortman K, Carbone FR, and Heath WR
- Subjects
- Animals, Autoantigens immunology, Autoantigens metabolism, CD36 Antigens genetics, CD36 Antigens physiology, CD8-Positive T-Lymphocytes immunology, Cell Death immunology, Cells, Cultured, Clonal Deletion immunology, Injections, Intravenous, Lymphocyte Transfusion, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Ovalbumin administration & dosage, Ovalbumin immunology, Phagocytosis immunology, Spleen immunology, Spleen metabolism, Spleen transplantation, Antigen Presentation, CD36 Antigens biosynthesis, CD8 Antigens biosynthesis, Dendritic Cells immunology, Dendritic Cells metabolism, Spleen cytology
- Abstract
Cross-presentation allows the processing of Ags from donor cells into the MHC class I presentation pathway of dendritic cells (DCs). This is important for the generation of cytotoxic T cell immunity and for induction of self tolerance. Apoptotic cells are reported to be efficient targets for cross-presentation, and in vitro studies using human DCs have implicated CD36 in their capture. In support of a role for CD36 in cross-presentation, we show that this molecule is differentially expressed by CD8(+) splenic DCs, which previously have been identified as responsible for cross-presentation in the mouse. Three different cross-presentation models were examined for their dependence on CD36. These included cross-priming to OVA-coated spleen cells and cross-tolerance to OVA transgenically expressed in the pancreatic islet beta cells under constitutive conditions or during beta cell destruction. In these models, CD36 knockout DCs were equivalent to wild-type DCs in their capacity to cross-present either foreign or self Ags, indicating that CD36 is not essential for cross-presentation of cellular Ags in vivo.
- Published
- 2002
- Full Text
- View/download PDF
40. The cross-priming APC requires a Rel-dependent signal to induce CTL.
- Author
-
Mintern JD, Belz G, Gerondakis S, Carbone FR, and Heath WR
- Subjects
- Animals, Antigen Presentation genetics, Antigen-Presenting Cells cytology, Antigens, Viral administration & dosage, CD40 Antigens physiology, Cell Differentiation genetics, Cell Differentiation immunology, Epitopes, T-Lymphocyte immunology, Herpesvirus 1, Human immunology, Injections, Intravenous, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Ovalbumin immunology, Proto-Oncogene Proteins c-rel biosynthesis, Proto-Oncogene Proteins c-rel deficiency, Proto-Oncogene Proteins c-rel genetics, Signal Transduction genetics, Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, Cytotoxicity, Immunologic genetics, Lymphocyte Activation genetics, Proto-Oncogene Proteins c-rel physiology, Signal Transduction immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
Induction of OVA-specific CTL by cross-priming requires help from CD4 T cells, which use CD154 to signal CD40 on the APC. To further dissect the molecular pathways involved in cross-priming, we examined the role of Rel, an NF-kappaB family member. c-rel(-/-) mice failed to generate OVA-specific CTL by cross-priming, but could induce CTL to HSV-1. Using chimeric mice, Rel expression was shown to be required by the APC, but not by the T cells. Notably, the deficiency in Rel could be overcome by triggering CD40, implying that the APC required Rel before receipt of the CD40 signal. These data suggest that the cross-priming APC must receive two signals before it can stimulate CTL. The first signal is Rel dependent and is required before activation of CD4 helper T cells, which then deliver the second signal using CD154 to trigger CD40.
- Published
- 2002
- Full Text
- View/download PDF
41. Cutting edge: precursor frequency affects the helper dependence of cytotoxic T cells.
- Author
-
Mintern JD, Davey GM, Belz GT, Carbone FR, and Heath WR
- Subjects
- Animals, Antigens pharmacology, CD40 Ligand biosynthesis, CD40 Ligand physiology, Epitopes, T-Lymphocyte administration & dosage, Epitopes, T-Lymphocyte immunology, Histocompatibility Antigens Class II genetics, Injections, Intravenous, Lymphocyte Activation genetics, Lymphocyte Count, Mice, Mice, Inbred C57BL, Mice, Transgenic, Ovalbumin administration & dosage, Ovalbumin genetics, Ovalbumin immunology, Spleen cytology, Spleen immunology, Spleen transplantation, T-Lymphocytes, Cytotoxic transplantation, Stem Cells cytology, Stem Cells immunology, T-Lymphocytes, Cytotoxic cytology, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Helper-Inducer immunology
- Abstract
Generation of CTL immunity often depends on the availability of CD4 T cell help. In this report, we show that CTL responses induced by cross-priming can be converted from CD4-dependent to CD4-independent by increasing the frequency of CTL precursors. In the absence of CD4 T cells, high numbers of CTL precursors were able to expand in number and become effector CTL. The ability of high frequencies of CD8 T cells to override help was not due to their ability to signal CD40 via expression of CD154. These findings suggest that when precursor frequencies are high, priming of CD8 T cell responses may not require CD4 T cell help.
- Published
- 2002
- Full Text
- View/download PDF
42. Progression of armed CTL from draining lymph node to spleen shortly after localized infection with herpes simplex virus 1.
- Author
-
Coles RM, Mueller SN, Heath WR, Carbone FR, and Brooks AG
- Subjects
- Animals, Cell Division immunology, Epitopes, T-Lymphocyte analysis, Herpes Simplex pathology, Hindlimb, Histocompatibility Antigens Class I analysis, Injections, Subcutaneous, Lymph Nodes pathology, Lymphocyte Activation, Lymphoid Tissue immunology, Lymphoid Tissue pathology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Spleen pathology, Viral Envelope Proteins immunology, Cell Movement immunology, Cytotoxicity, Immunologic, Herpes Simplex immunology, Herpesvirus 1, Human immunology, Lymph Nodes immunology, Spleen immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
We have examined the generation of CTL immunity immediately after localized footpad infection with herpes simplex virus 1 (HSV-1) using three coordinated in vivo T cell tracking methodologies. Tetrameric MHC class I containing the immunodominant peptide from HSV-1 glycoprotein B (gB) showed that after infection the proportion of Ag-specific T cells peaked at day 5 within draining popliteal lymph nodes and 2 days later in the spleen. Preferential expression of the activation marker CD25 by tetramer-positive cells in draining popliteal nodes but not spleen suggested that gB-specific T cells were initially activated within the lymph node. In vivo cytotoxicity assays showed that Ag-specific effector cells were present within the draining lymph nodes as early as day 2 after infection, with a further 2-day lag before detection in the spleen. Consistent with the very early arming of effector CTL in the draining lymph node, adoptive transfer of CFSE-labeled gB-specific transgenic T cells showed that they had undergone one to four rounds of cell division by day 2 after infection. In contrast, proliferating T cells were first detected in appreciable numbers in the spleen on day 4, at which time they had undergone extensive cell division. These data demonstrate that HSV-1-specific T cells are rapidly activated and armed within draining lymph nodes shortly after localized HSV-1 infection. This is followed by their dissemination to other compartments such as the spleen, where they further proliferate in an Ag-independent fashion.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.